CLEC4G has dual roles in immune modulation and neurodegeneration:
Inhibits T-cell activation via BTN3A1 binding, reducing cytokine production (IFN-γ, IL-2, TNF-α) and chemokines (CXCL9, CCL17) .
Acts as a co-inhibitory checkpoint in melanoma, suggesting therapeutic potential .
Reduces Aβ40 in neurons by interacting with BACE1, a key enzyme in amyloid-beta production .
Expression inversely correlates with Alzheimer’s disease (AD) progression:
CLEC4G (C-type lectin domain family 4 member G), also known as LSECtin, is a type II transmembrane glycoprotein approximately 40 kDa in size and 293 amino acids in length. It belongs to the C-type (Ca²⁺-dependent) lectin superfamily and contains:
A short N-terminal cytoplasmic tail (amino acids 1-31)
A 21 amino acid transmembrane region
An extracellular region with:
Two N-linked glycosylation sites (amino acids 73 and 159)
A coil-coil neck domain (amino acids 96-136)
A C-type lectin-like domain (CTLD) (amino acids 165-289)
A C-terminal Ca²⁺-dependent carbohydrate-recognition domain (C-type CRD)
CLEC4G is significant in research due to its roles in:
Glycan binding (mannose, GlcNAc, and fucose) in a Ca²⁺-dependent manner
Viral recognition and binding (SARS coronavirus, Ebola virus, Japanese encephalitis virus)
Potential neuroprotection against Alzheimer's disease progression through inhibition of Aβ generation
CLEC4G expression has been identified in multiple tissues:
When selecting antibodies, consider:
For liver studies: Antibodies targeting membrane epitopes may be more effective
For neuronal studies: Antibodies recognizing monomeric forms and cytoplasmic epitopes are preferable
For experimental validation: Use appropriate positive control tissues (liver for high expression, neuronal tissues for cytoplasmic localization)
Flow Cytometry:
Harvest cells (dendritic cells, transfected cell lines)
Wash with PBS containing 0.5% BSA
Block Fc receptors if working with immune cells
Stain with anti-CLEC4G antibody (e.g., Mouse Anti-Human LSECtin/CLEC4G Monoclonal Antibody, MAB2947 at manufacturer's recommended dilution)
Incubate for 30 minutes at 4°C
Wash twice
If using unconjugated primary antibody, stain with appropriate secondary (e.g., Allophycocyanin-conjugated Anti-Mouse IgG)
Immunohistochemistry:
Fix tissue sections (formalin-fixed paraffin-embedded or frozen)
For FFPE: Perform antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0
Block endogenous peroxidase and non-specific binding
Apply anti-CLEC4G antibody at recommended dilution (e.g., 1:500-1:2000 for polyclonal antibodies like 18173-1-AP)
Incubate overnight at 4°C
Apply appropriate detection system
Western Blotting:
Prepare protein lysates from tissues or transfected cells
Separate proteins by SDS-PAGE
Transfer to membrane
Block and incubate with anti-CLEC4G antibody (recommended dilution: 1:500-1:5000)
Expected band size: approximately 40 kDa
Note: Expression levels may require enrichment or use of transfected cells as positive controls
A robust validation strategy should include:
Positive and negative controls:
Multiple detection methods:
Blockade experiments:
Pre-incubate antibody with recombinant CLEC4G protein before staining
Signal should be significantly reduced or eliminated
siRNA knockdown:
Reduce CLEC4G expression in appropriate cell types
Compare antibody staining in knockdown versus control cells
Isotype controls:
Recent research has identified CLEC4G as having potential neuroprotective functions in Alzheimer's disease (AD). Antibody-based approaches to study this relationship include:
Expression analysis in AD progression:
Co-localization studies with AD markers:
Double-label immunofluorescence with CLEC4G and BACE1 antibodies
Design: Use confocal microscopy with spectrally distinct fluorophores
Controls: Include single antibody staining to control for bleed-through
Mechanistic investigation of CLEC4G-BACE1 interaction:
Co-immunoprecipitation with CLEC4G antibodies to pull down protein complexes
Western blot analysis to detect BACE1 in the precipitates
Validate with reciprocal co-IP using BACE1 antibodies
Functional analysis in neuronal models:
Age-dependent analysis in APP/PS1 mouse models:
When faced with conflicting results from different antibody clones:
Epitope mapping analysis:
Determine the binding sites of each antibody clone
Different epitopes may be differentially accessible depending on protein conformation or processing
Method: Use peptide arrays covering the CLEC4G sequence
Multiple detection methodologies:
Compare results across Western blot, immunoprecipitation, flow cytometry, and imaging
Discrepancies may be method-specific rather than antibody-specific
Splice variant analysis:
Post-translational modification considerations:
CLEC4G contains N-glycosylation sites that may affect antibody binding
Test with deglycosylated samples to determine if glycosylation impacts recognition
Independent validation strategies:
Use genetic approaches (CRISPR/Cas9, siRNA) to modulate CLEC4G expression
Employ orthogonal methods such as RNA-seq to correlate with protein detection
Consider mass spectrometry-based validation
CLEC4G functions as a receptor for several viruses, making it valuable for infectious disease research:
Viral binding studies:
Use CLEC4G antibodies to block viral attachment to cells
Methodology: Pre-incubate cells with blocking anti-CLEC4G antibodies before viral challenge
Readout: Measure infection rates in blocked versus unblocked cells
Mechanistic analysis of CLEC4G-viral interactions:
Immunoprecipitate CLEC4G with antibodies after viral exposure
Western blot for viral proteins in the precipitates
Mass spectrometry analysis of co-precipitated proteins
Identification of binding domains:
Use domain-specific CLEC4G antibodies to determine which regions are critical for viral binding
Compare antibodies targeting the CTLD versus other domains
Correlate binding inhibition with viral entry inhibition
Therapeutic antibody development:
Screen for antibodies that specifically block viral binding without disrupting normal CLEC4G function
Test antibody fragments (Fab, scFv) for improved tissue penetration
Evaluate humanized versions of effective blocking antibodies
Comparative analysis across viral families:
Several factors can explain variable staining patterns:
For co-localization studies with CLEC4G and interaction partners (e.g., BACE1):
Antibody compatibility:
Select primary antibodies from different host species (e.g., mouse anti-CLEC4G and rabbit anti-BACE1)
If same-species antibodies must be used, consider directly conjugated antibodies or sequential staining protocols
Signal optimization:
For weak CLEC4G signals in neuronal tissues, use tyramide signal amplification
Balance signal strengths by adjusting antibody concentrations individually before combining
Preventing cross-reactivity:
Include additional blocking steps between primary antibodies if using sequential protocols
Validate each antibody independently before combining
Include controls with each primary alone to check for cross-reactivity of secondaries
Proximity ligation assay (PLA):
For detecting protein-protein interactions within 40nm
Use species-specific PLA probes against CLEC4G and BACE1 antibodies
Quantify PLA signals in different brain regions or cell types
Confocal optimization:
Use spectral unmixing for fluorophores with overlapping emission spectra
Employ sequential scanning to prevent bleed-through
Include single-labeled controls on the same slide
For quantitative analysis of CLEC4G in disease contexts:
Standardization protocols:
Use recombinant CLEC4G protein standards for calibration
Include the same positive control tissue in each experiment
Apply batch correction methods for samples processed at different times
Quantification methods:
For IHC/IF: Use digital pathology software with validated algorithms for intensity measurement
For flow cytometry: Include calibration beads to normalize mean fluorescence intensity
For Western blot: Use housekeeping proteins appropriate for the tissue/condition
Age and disease stage considerations:
Statistical approaches:
Use appropriate statistical tests based on data distribution
Account for potential confounding variables (age, sex, postmortem interval)
Calculate minimum sample sizes needed for desired statistical power
Validation across methodologies:
Confirm protein-level changes with transcript-level measurements
Consider single-cell approaches to account for cellular heterogeneity
Use orthogonal methods (e.g., ELISA and Western blotting) to validate quantitative changes
Single-cell analysis using CLEC4G antibodies can reveal:
Cell-type heterogeneity:
Developmental regulation:
Disease-specific changes:
Spatial context:
Combine with spatial transcriptomics or multiplexed imaging
Map CLEC4G expression patterns in relation to brain regions and pathological features
Identify microenvironmental factors influencing expression
CLEC4G's role in glycan binding suggests connections to neurodegeneration:
N-glycan interactions:
Correlation with glycosylation enzymes:
Functional implications:
Therapeutic potential:
CLEC4G's ability to recognize specific glycan structures could be leveraged for targeted interventions
Approach: Screen for compounds that enhance CLEC4G-mediated clearance of pathological glycans
Recent research has identified key transcriptional regulators of CLEC4G:
Upstream transcription factors:
Regulatory network differences:
Modulation strategies:
Target NR2F6 and XRCC4 to potentially upregulate CLEC4G expression
Experimental design: Overexpress these factors in neuronal cells and measure CLEC4G levels
Validation: Use CLEC4G antibodies to quantify protein changes following transcription factor modulation
Epigenetic considerations:
Investigate methylation status of CLEC4G promoter in different brain regions
Combine with antibody-based detection to correlate epigenetic marks with protein expression
Approach: Use methylation-specific antibodies alongside CLEC4G antibodies